Skip to main content

ORIGINAL RESEARCH article

Front. Pharmacol., 08 January 2018
Sec. Ethnopharmacology

The Calcium-Induced Regulation in the Molecular and Transcriptional Circuitry of Human Inflammatory Response and Autoimmunity

  • State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China

Rheumatoid arthritis synovial fibroblasts (RASFs) are fundamental effector cells in RA driving the joint inflammation and deformities. Celastrol is a natural compound that exhibits a potent anti-arthritic effect promoting endoplasmic reticulum (ER) stress mediated by intracellular calcium (Ca2+) mobilization. Ca2+ is a second messenger regulating a variety of cellular processes. We hypothesized that the compound, celastrol, affecting cytosolic Ca2+ mobilization could serve as a novel strategy to combat RA. To address this issue, celastrol was used as a molecular tool to assay the inflammatory gene expression profile regulated by Ca2+. We confirmed that celastrol treatment mobilized cytosolic Ca2+ in patient-derived RASFs. It was found that 23 genes out of 370 were manipulated by Ca2+ mobilization using an inflammatory and autoimmunity PCR array following independent quantitative PCR validation. Most of the identified genes were downregulated and categorized into five groups corresponding to their cellular responses participating in RA pathogenesis. Accordingly, a signaling network map demonstrating the possible molecular circuitry connecting the functions of the products of these genes was generated based on literature review. In addition, a bioinformatics analysis revealed that celastrol-induced Ca2+ mobilization gene expression profile showed a novel mode of action compared with three FDA-approved rheumatic drugs (methotrexate, rituximab and tocilizumab). To the best of our knowledge, this is a pioneer work charting the Ca2+ signaling network on the regulation of RA-associated inflammatory gene expression.

Introduction

Rheumatoid arthritis (RA) is the most common chronic systemic autoimmune inflammatory disease. RA affects around 1% of people worldwide, and is more prevalent in women (Bartok and Firestein, 2010; Burmester and Pope, 2017). The quality of life in RA patients is critically compromised, which usually causes progressive articular destruction, early unemployment, and considerable disability implying a huge socioeconomic burden (Albers et al., 1999; Russell, 2008). RA is characterized by synovitis associated with the formation of a hyperplastic synovial membrane, which contains large number of immunocellular components including T and B cells, plasma cells, mast cells, macrophages, and activated RA synovial fibroblasts (RASFs) (Bartok and Firestein, 2010). The cellular interplay between these cells transforms the synovium into an invasive pannus and promotes angiogenesis, as well as the release of cytokines, chemokines, and matrix-degrading enzymes which facilitates the development of joint tissue damage. In particular, the RASFs, which actively proliferate and develop resistance to apoptosis are the major cell population in the synovial lesion (Perlman et al., 2001; Firestein, 2003; Muller-Ladner et al., 2007; Bartok and Firestein, 2010). These factors, coupled with activated RASFs' capability of secreting the aforementioned mediators of RA, establishes a paracrine/autocrine cycle that perpetuates RA synovitis, recruitment of new cells to the affected joint and induces joint destruction (Bartok and Firestein, 2010), which represents the potential therapeutic target of RASFs for achieving long-term remission of RA.

Early diagnosis and treatment is critical to the diminution of the seriousness of RA. Currently, non-steroidal anti-inflammatory drugs (NSAIDs), glucocorticoids (steroid hormones), and disease-modifying antirheumatic drugs (DMARDs) are standard pharmaceutical interventions for the inflammatory disorder. Of note, DMARDs medications suppress immune system functions to prevent further structural damage of bone and cartilage in the affected joints (Burmester and Pope, 2017; McInnes and Schett, 2017). Amongst which methotrexate (MTX) is the first-line therapy commonly prescribed, and the addition of targeted DMARD, such as tumor necrosis factor (TNF)-inhibitors, interleukin-6-inhibitors, B-cell depleting drugs, and Janus kinase (JAK)-inhibitors, are adjuvant interventions when poor prognostic factors are present. These new treat-to-target medications have improved the course of RA and most of them modulate specific mechanistic steps in the inflammatory process. However, a considerable numbers of individuals do not respond adequately to or are suffering from side-effects toward such treatments (Burmester and Pope, 2017; Tarp et al., 2017). Therefore, a better understanding of the molecular machinery associated with RA pathogenesis and the complex crosstalk between the constituting signaling pathways might help to facilitate the development or improvement of novel and conventional therapeutic strategies.

The Chinese medicinal herb Tripterygium wilfordii Hook f (TwHF) is a traditional remedy for RA treatment (Tao et al., 2002; Tang and Zuo, 2012). In fact, the recent randomized clinical trial studies further confirmed the therapeutic efficacy of TwHF in patients with active RA (Lv et al., 2015). Celastrol is the bioactive ingredient constituting TwHF which has demonstrated anti-proliferative and anti-inflammatory properties in both in vitro and in vivo models (Brinker et al., 2007; Kim et al., 2009; Venkatesha et al., 2011; Cascão et al., 2012; Nanjundaiah et al., 2012). It has been shown that celastrol promotes endoplasmic reticulum (ER) stress mediated by intracellular calcium (Ca2+) mobilization (Yoon et al., 2014). Ca2+ as a second messenger is required for the regulation of many cellular processes, including gene transcription, cell shape, motility, proliferation, mitochondrial function, apoptosis, and immune responses (Clapham, 2007). As early as two decades ago, the role of cellular Ca2+ in diseases was being noticed, from cardiovascular diseases to strokes, diabetes, the immune response (including inflammation) and cancer (Mooren and Kinne, 1998). More recently, studies have shown that intracellular Ca2+ signaling has been implicated in the pathogenesis of autoimmune disorders, such as RA (Izquierdo et al., 2014) and that altered cellular Ca2+ homeostasis is related to the control of various hallmarks of cancer. (Marchi and Pinton, 2016). Modulation of Ca2+ signaling has even been proposed as a possible therapy option for the treatment of cancer, though this remains as yet relatively unexplored (Rooke, 2014). Accordingly, celastrol is a suitable tool for investigating the role of Ca2+ signaling in pathomechanisms of RA. In this report, we hypothesized that compounds affecting cytosolic Ca2+ mobilization might serve to combat RA disease. The inflammatory gene expression profile which are regulated by Ca2+ dynamic changes in response to celastrol treatment were identified in patient-derived RASF. In addition, the possible transcriptional and molecular circuitry associated with the cytosolic Ca2+ mobilization was unraveled through literature review and bioinformatics analysis. Our findings provide novel insight into the application of ion channel modulators for RA intervention.

Materials and Methods

Isolation and Culture of RASF

Primary culture RASF were isolated from synovium obtained from RA patients who had undergone knee surgery for synovectomy. Diagnosis of RA in these patients was made according to the American Rheumatism Association's 1987 revised criteria for classification of RA (Arnett et al., 1988). Synovial strips were cut into small pieces, placed in a 25 cm2 culture flask, and then cultured in DMEM containing 20% fetal bovine serum (FBS). Medium was changed every 3 days and, after 2 weeks, the synovial tissues were removed from the cultured medium. RASFs were digested by 0.25% trypsin for 5 min at 37°C. The cell suspension was diluted with DMEM containing 20% FBS and 1% Penicillin/Streptomycin with L-Glutamine (PSG), and separated into other flasks. Cell cultures were maintained at 37°C in a humidified incubator (atmosphere of 5% CO2). The purification of RASFs was validated by staining CD90. Cultured RASFs from passages 5–7 were employed for the below analysis. Unless otherwise specified, all reagents were purchased from Sigma-Aldrich (MO, USA).

Measurement of Cytoplasmic Calcium Dynamic

Intracellular cytosolic Ca2+ dynamic was measured using the FLIPR Calcium 6 Assay Kit (Molecular Devices, USA), which contains a proprietary Ca2+-sensitive fluorophore, according to the manufacturer's instructions. In brief, 10000 RASFs per well were seeded in black wall/clear bottom 96-multiwell plates from Costar (Tewksbury, MA, USA) and cultured for 24 h before treatment. After that, calcium 6 reagent was added directly to cells, and cells were incubated for an additional 2 h at 37°C and 5% CO2. One micromolar of celastrol (China Chengdu MUST, A000106) was then added to the wells and immediately subjected to data acquisition on the FLIPR Tetra High-Throughput Cellular Screening System (Molecular Devices, USA) at room temperature using a 1-s reading interval throughout the experiments.

Single Cell Calcium Imaging

2 × 105 RASFs cells were cultured in 35 mm confocal disc at 37°C CO2 incubator for 24 h. FLIPR Calcium 6 reagent was added to cells at 37°C for 30 min. RASFs were then washed 3 times with HEPES buffer saline and incubated at 37°C in an imaging chamber for another 10 min. Changes in cytosolic [Ca2+] levels were monitored by following changes in FLIPR Calcium 6 fluorescence upon addition of 1 μM celastrol in HBSS buffer, using the real-time mode for 4 min by epifluorescence microscopy (Applied Precision DeltaVision Elite, Applied Precision, Inc., USA). Data Inspection Program provided by the DeltaVision software was used to measure the intensity of the FLIPR Calcium 6 fluorescence and the mean fluorescence intensity was monitored at 525 nm and plotted against time (s).

RNA Extraction & cDNA Synthesis

RNA was extracted using RNeasy Mini Kit (Qiagen, USA) from RASFs untreated (control), treated with celastrol [1 μM], or celastrol in the presence of BAPTA/AM [10 μM] (Santa Cruz, USA) for 24 h. RNA concentration was determined using the NanoDrop 2000c Spectrophotometer (Thermo Scientific) and 1 μg of RNA was used to synthesize cDNA with RT2 First Strand Kit (Qiagen, USA). Three independent biological samples were employed.

RT2 Profiler™ PCR Array–Inflammatory Gene Expression Profiling

Quantitative PCR was performed using the cDNA prepared from RASFs with RT2 SYBR® Green qPCR Mastermix (Qiagen, USA) and ViiA™ 7 Real Time PCR System (Applied Biosystems). The RT2 Profiler™ PCR Array Human Inflammatory Response & Autoimmunity 384HT kit (Qiagen, USA) was assessed according to the manufacturer's instructions. This PCR array contains primers for 370 genes related to various functions of the immune system, from cytokines/chemokines to their receptors, cytokine production, and other proteins and receptors involved in acute-phase, inflammatory and humoral immune responses. Analysis of results was achieved using the integrated web-based RT2 Profiler™ PCR Array Data Analysis software from Qiagen, which calculated all ΔΔCt-based fold-change and fold-regulation from three independent raw data. Validation of the identified gene expression pattern upon celastrol-induced calcium mobilization was performed by quantitative PCR with self-designed primers (Tech Dragon Ltd., Hong Kong). Primer sequences (see Supplementary Table) were designed employing ThermoFisher Scientific's online OligoPerfect™ Designer software and then verified with NCBI's Primer-BLAST software to confirm specific recognition of target genes. Gene expressions were normalized to GAPDH, relative to control, and analyzed using the 2−ΔΔCT method. All the data were statistically analyzed by unpaired t-test.

Bioinformatics Analysis

The identified celastrol-mediated Ca2+ mobilized genes in this report were searched for their implication in Ca2+-dependent function using NCBI database and KEGG pathways. Furthermore, NCBI database (Geo DataSets) was searched for datasets that were generated by treating RA patient with FDA-approved drugs. Three studies were selected that evaluated Rituximab (GDS4903), Tocilizumab (GDS5068), and Methotrexate (GDS5069) for RA treatment. The gene expression profile was transformed into fold change (2−ΔΔCT method) and merged with our dataset (celastrol-mediated Ca2+ mobilization regulated genes) using R (3.3.2). Merged datasets was then processed with Phyloseq (1.19.1) and ggplot2 (2.2.1) packages to generate principal coordinate plot with weighted UniFrac distance. Additionally, SIMPER analysis was performed with R (3.3.2) using vegan (2.4-3) to determine genes that are similarly responding to celastrol [Ca2+] and FDA-approved drugs (Rituximab, Tocilizumab, and Methotrexate).

Results

Celastrol Modulated Inflammatory and Immunity Genes via Ca2+ Mobilization

To confirm the effect of celastrol on Ca2+ mobilization in RASFs, intracellular cytosolic Ca2+ dynamic and single live-cell Ca2+ imaging was performed on celastrol-stimulated RASFs. As shown in Figures 1A,B and Supplementary Video, RASFs loaded with FLIPR Calcium 6 displayed a dramatic increase in fluorescence intensity upon 1 μM of celastrol treatment. Celastrol induced Ca2+ dynamic changes in RASFs within c.a. 30 s, confirming that celastrol is a suitable tool for Ca2+-flux effects studies. Quantitative PCR was performed to identify the inflammatory genes expression profile affected by celastrol-mediated Ca2+ mobilization with the use of RT2 Profiler™ PCR Array Human Inflammatory Response & Autoimmunity. Only those genes with changes in fold regulation above ±1.5-fold in the PCR array result were considered to analyse in this study. After celastrol treatment, the expression of 134 out of the total 370 examined genes were up-regulated or down-regulated Figure 1C. Since, the exact mechanistic regulations that mediate such genes expression are still elusive, we investigated the potential role of Ca2+ in the expression of these genes. In the presence of Ca2+ chelator, BAPTA/AM, we identified that 72 out of the 134 genes were regulated by celastrol-mediated Ca2+ mobilization. In addition, those genes which commenced their expression at Ct ≥ 30 were excluded decreasing the number of validated genes to 40 Figure 1D. In order to confirm the reliability of the PCR array result, these 40 genes were further individually validated by quantitative PCR using in-house designed primers. We found that the expression profiles (measured in fold change instead of fold regulation) of 23 out of the 40 genes are consistent with the data observed in the PCR array (Table 1). Of note, celastrol downregulated almost all of the genes (22 of them) under examination in a cytosolic Ca2+-dependent manner except CD40 which was upregulated in response to the changes in cellular Ca2+ level (Figure 2).

FIGURE 1
www.frontiersin.org

Figure 1. (A) Celastrol induced calcium dynamic change in RASFs. Cells treated with 1 μM celastrol were loaded with FLIPR Calcium 6 dye. Real time Ca2+ kinetic was monitored with FLIPR Tetra instrument. Data from the chart represent mean values ± SD. of three independent experiments. (B) Single cell imaging visualized celastrol-mobilized cytosolic calcium level in RASFs. Cells treated with 1 μM celastrol were loaded with FLIPR Calcium 6 dye. Calcium signal was monitored by Applied Precision DeltaVision Elite in real-time mode (see Supplementary Video). Chart represents the mean intensity of fluorescence signal at 525 nm. (C) Scatter plot for inflammatory and immunity genes fold regulation values from celastrol stimulated RASFs relative to unstimulated RASFs (Control): genes not regulated (black), up-regulated genes (red), and down-regulated genes (green) with threshold lines of 1.5 and −1.5. (D) Scatter plot for the genes identified as up-regulated or down-regulated with celastrol treatment (in C). Dots represent the genes fold regulation values from RASFs treated with celastrol and BAPTA/AM relative to untreated control: genes not regulated (black), up-regulated genes (red), and down-regulated genes (green) with threshold lines of 1.5 and −1.5 (Ct < 30). Data from the scatter plots represent mean values of three independent experiments.

TABLE 1
www.frontiersin.org

Table 1. Gene expression (fold change relative to untreated control) regulated by celastrol-mediated Ca2+ mobilization analysis in RASFs.

FIGURE 2
www.frontiersin.org

Figure 2. Gene expression regulated by celastrol-mediated Ca2+ mobilization analysis in RASF. RT-qPCR independent validation from RASF cells untreated (control), or treated with 1 μM Celastrol (Cel), and 10 μM BAPTA/AM (BM) for 24 h. Gene expressions were normalized to GAPDH, relative to control, and analyzed using the 2−ΔΔCT method. The data is represented as the mean ± SD. **P ≤ 0.01; ***P ≤ 0.001 compared with control. #P ≤ 0.05; ##P ≤ 0.01; ###P ≤ 0.001 compared with Celastrol.

Ca2+ Modulated Genes Expression in RASFs Were Categorized into Five RA Pathogenic Factors

To illustrate the functional roles of the identified Ca2+-modulated genes, an intensive literature review was performed. These genes were further categorized into five groups according to their cellular functions as: “Apoptosis/cell death,” “Cell proliferation,” “Cell migration/invasion,” “Angiogenesis,” and “Immunity/inflammation” for analysis (Table 2). Such cellular processes are the key factors etiologically associated with the progress of RA inflammation. The aberrant proliferation (Firestein, 2003) and hampered apoptotic machinery (Perlman et al., 2001) of RASFs are responsible for pannus formation. The formation and progressive invasion of pannus, which consist of mainly RASFs, in RA joints are responsible for the bone and cartilage destruction and supported by extensive vascular overgrowth (Bartok and Firestein, 2010). Also, the spread of the symptoms from the affected site to different joints is related to the abnormal migratory ability of RASFs (Lefèvre et al., 2009). The release of different cytokines and chemokines are critical to attract the circulatory immunocellular components infiltrating the RA-affected synovium (Bartok and Firestein, 2010). The 23 identified genes appeared to be multi-functional and manipulate a multitude of physiological responses of RASFs. For example, the regulatory effects of bone morphogenetic protein 1 (BMP1), calpastatin (CAST), and Toll-like receptor 6 (TLR6), participate in all of the above described cellular functions. Among these genes, 17 of them play a role in apoptosis/cell death, 17 genes regulate cell migration/invasion, 15 genes are involved in angiogenesis, 22 genes can promote or inhibit cell proliferation, and 21 genes are associated with immunity/inflammation. The products of these genes represent a great variety ranging from upstream cellular receptors, e.g., TLR6 and leptin receptor (LEPR), signaling molecules like cytokines and enzymes, e.g., IK cytokines (IK) and BMP1, to downstream transcriptional factors, e.g., nuclear factor of activated T-cells c3 (NFATC3) and signal transducer and activator of transcription 3 (STAT3). In addition, apart from the genes CAST, CD40, NFATC3, and TRAP1, which had previously been found to be regulated by Ca2+-signaling machinery (Casanova et al., 2006; Hanna et al., 2008; Landriscina et al., 2010; Brun and Godbout, 2016), the remaining 19 genes were newly discovered to have their expression regulated by cytosolic Ca2+.

TABLE 2
www.frontiersin.org

Table 2. Association of the validated calcium-modulated genes with five important RA pathogenesis factors. Genes were categorized by pathogenic factors (enhancement or inhibition) through the use of NCBI database.

Perspective Signaling Network Connecting the Ca2+-Modulated Inflammatory Genes in RA

Accordingly, a network scheme (Figure 3) involving the identified genes (yellow boxes), as well as the other related genes and pathways were mapped to demonstrate the Ca2+ involvement in RA pathogenesis. The network map illustrated major Ca2+-signaling pathways: (1) calpastatin (CAST)/calpains (Minobe et al., 2006; Hanna et al., 2008) and (2) calpains/calmodulin-calcineurin-NFATc3 axes (Hernández et al., 2001; Dai et al., 2005; Lee et al., 2011; Li et al., 2011; Neria et al., 2013; Yoon et al., 2013; Jia et al., 2014; Baron et al., 2015; Kar and Parekh, 2015; Brun and Godbout, 2016; Mognol et al., 2016) (blue boxes and arrows). Some pathways that are positively regulated by calpains (orange arrows) are well-known pathways such as NF-κB signaling axis (Li et al., 2011, 2014; Storr et al., 2011, 2015) (green boxes and arrows; includes NFKB1, which encodes the precursor protein of p50, one of the subunits of NF-κB, Karin and Ben-Neriah, 2000), JAK/STAT axis (purple boxes and arrows; includes STAT3) (Pothlichet et al., 2008; Miyazaki et al., 2015), and the ERK1/2 axis (Moshal et al., 2006) (pale red boxes and arrows). Calpain has been demonstrated to inhibit two apoptotic pathways by cleaving p53 (Storr et al., 2015) and Myc (Niapour et al., 2008; Storr et al., 2011). When released to the extracellular medium, it also promotes proliferation by converting Cyclin E into a hyperactive form (Storr et al., 2011) which may directly break down cartilage (Ishikawa et al., 1999). Another protein, TRAP1, which is stabilized by the Ca2+-dependent protein Sorcin, promotes cell proliferation and inhibits apoptosis (Landriscina et al., 2010). The other identified calcium-modulated genes have also been found to activate pathways such as: JAK/STAT axis [IFNAR1 (Walters and Jelinek, 2004; Qian et al., 2011), LEPR (Sanchez-Margalet and Martin-Romero, 2001), and TRAP1 (Ou et al., 2014)]; PI3K/Akt axis [ERBB2 (Woods Ignatoski et al., 2003), IL1R1 (Sizemore et al., 1999), IL4R (Dubois et al., 1998), LEPR (Uddin et al., 2010), and FGF10 (Li et al., 2016)] and PI3K/PKC/NF-κB axis [green boxes and arrows; HRH1, (Dickenson, 2002) and ERBB2 (Woods Ignatoski et al., 2003)]; and also NF-κB signaling pathway through other signaling cascades [CD40 (Lee et al., 2006), IFNAR1 (Yang et al., 2008), IL1R1 (Dower et al., 1986; Burns et al., 1998; Ahmad et al., 2007), and TLR6 (de Almeida et al., 2013)]; and MyD88-dependent MAPKs/AP-1 axis [light blue boxes and arrows; activated by IL1R1 (Dower et al., 1986; Burns et al., 1998; Ahmad et al., 2007), TLR6 (de Almeida et al., 2013), and NFRKB (Audard et al., 2012)]. The identified gene TOLLIP inhibits IL1R1 and TLR6 function (Burns et al., 2000; Bulut et al., 2001), and thereby prevents the activation of MyD88-dependent MAPK/AP-1 and NF-κB pathways, as well as PI3K pathways. GLMN, encoding the protein glomulin, also known as FAP48 or FAP68, has been shown to upregulate IL-2 expression (Krummrei et al., 2003). Other effects caused by activation of these mentioned genes include ERBB2 upregulating CAST expression (Ai et al., 2013) and HRH1 upregulating MMP1, possibly through AP-1 activation (Zenmyo et al., 1995). The other identified genes including ADGRE5, BMP1, CMTM1, IK, NFX1, and SCUBE1 are still elusive in an inflammatory pathways. In summary, the network presented in Figure 3 revealed the possible connection between the Ca2+-signaling axis and the identified calcium-modulated genes.

FIGURE 3
www.frontiersin.org

Figure 3. Network map linking some of the Ca2+-modulated genes to Ca2+signaling pathways and five RA pathogenic factors. Literature review showing only the enhanced effects that drive RA pathogenesis.

Mode of Action of Celastrol-Induced Ca2+-Mobilization (Celastrol-[Ca2+]) Was Unique in Modulating Gene Expression Compared with the FDA-Approved RA Drugs

In order to determine whether the mode of action of celastrol-[Ca2+] gene expression is novel mechanism for RA management, bioinformatics analysis was employed to compare the results obtained in this study with those from FDA-approved drugs. Three datasets were downloaded from NCBI database that were comprised of genes expression profile (before and after treatment) of RA patients with methotrexate, rituximab, and tocilizumab. The datasets were subset to the list of genes that matched with celastrol-[Ca2+] regulated genes and comparatively analyzed using UniFrac distance analysis. As shown in Figure 4A, celastrol-[Ca2+] differentially modulated the genes expression. However, celastrol-[Ca2+] regulated genes were comparable with changes observed after methotrexate and tocilizumab treatments, whereas, greater dissimilarity was observed between rituximab and celastrol-[Ca2+]. Moreover, the genes that responded similarly to celastrol-[Ca2+] and methotrexate (Figure 4B), celastrol-[Ca2+] and rituximab (Figure 4C), and celastrol-[Ca2+] and tocilizumab (Figure 4D) were determined using SIMPER analysis using Bray-Curtis method. These findings suggested that the celastrol-[Ca2+] associated anti-inflammatory mechanism could be a specific target for RA intervention.

FIGURE 4
www.frontiersin.org

Figure 4. Comparative analysis of celastrol-mediated Ca2+ mobilization (celastrol-[Ca2+]) regulated genes with FDA-approved drug for RA. (A) Principal coordinates analysis with weighted UniFrac distance. Axes are showing percentile variations among the groups. Each dot corresponds to studied sample and respectively colored to sample types. (B) SIMPER analysis of the selected genes expression profile that are regulated by celastrol-[Ca2+] and methotrexate. (C) SIMPER analysis of the selected genes expression profile that are regulated by celastrol-[Ca2+] and rituximab. (D) SIMPER analysis of the selected genes expression profile that are regulated by celastrol-[Ca2+] and tocilizumab. X-axis is showing percentile similarity of the genes between the compared groups. The genes with higher percentage are more similar in responses toward celastrol-[Ca2+] and respective FDA-approved drug. The fold change of each gene after drug treatment has been shown in heat map.

Discussion

In this study, we found that celastrol significantly downregulated the expression of a number of genes related to the control of RASFs cellular pathophysiology. In fact, the suppressive effects of celastrol in immunoregulatory genes expression have been documented. By using multiplex analysis, Venkatesha et al. demonstrated that celastrol can reduce the expression of cytokines, such as TNF-α and IL-1β, and chemokines including MCP-1, MIP-1α, and RANTES, in different immunocellular components collected from a murine adjuvant-induced arthritis (AIA) model (Venkatesha et al., 2012). However, the detail molecular linkages between celastrol and such inflammatory-associated signaling are yet to be defined. Our PCR array and quantitative PCR data validated that the expression of a significant number (close to 20%) of these inflammatory genes was manipulated by celastrol in a Ca2+-dependent manner. Most importantly, the expression of these genes, critical to the perpetuation of the inflammatory phenotype of RA, were sensitive toward cytosolic Ca2+ accumulation as illustrated by the BAPTA/AM treatment suggesting the efficacy of targeting cellular Ca2+ level in RA therapy. Intriguingly, CD40 receptor was the only upregulated gene upon Ca2+ mobilization amongst the reported genes. In fact, the engagement of CD40 ligand and RASFs-expressed CD40 receptor can activate RANKL induction via ERK-1/2, p38 MAPK, and NF-κB activation and result in osteoclast hyperplasia which leads eventually to cartilage and bone destruction (Lee et al., 2006). Such observation further suggested the significant role of cellular Ca2+ in RA pathogenesis, which also aroused the concern of adverse effects associated with celastrol, since CD40 upregulation may activate the downstream inflammatory pathways worsening the RA progression. However, as demonstrated in our network map, such inflammatory pathways could, at the same time, be downregulated by other celastrol-regulated genes. On the other hand, celastrol induced downregulation of LEPR, cognate receptor for the satiety factor leptin, implying the possibility of abnormal weight gain of animals or patients upon celastrol treatment. Recently, Liu et al., found that prolonged exposure of mice with celastrol lead eventually to weight loss of the animals without inducing any toxicity and that mice with desensitized leptin signaling phenotype presented no significant responses to celastrol in terms of their weight (Liu et al., 2015). Therefore, it is reasonable to conclude that administration of celastrol in a reasonable dosage will not create significant toxicity which suggested the therapeutic value of our findings. In fact celastrol treatment with the use of AIA rat model has been reported to mitigate inflammatory RA with no toxicity, including the liver and kidney damages, demonstrated (Cascão et al., 2015).

Many receptors and transporter proteins are responsible for maintaining cellular Ca2+ homeostasis (Schwaller, 2012), for example, sarco/endoplasmic reticulum Ca2+-ATPase (SERCA), inositol trisphosphate receptor (InsP3), and ryanodine receptor (RyR) which located on the ER are the key managers of intracellular Ca2+ storage. In addition, the sodium-calcium (Liu et al., 2015) exchanger NCX and uniporter on the mitochondria are another pair of intracellular Ca2+ receptors important for cytosolic Ca2+ regulation. On the plasma membrane, the plasma membrane Ca2+ ATPase (PMCA), together with, other Ca2+ channels such as voltage-operated calcium channel (VOCC), receptor-operated calcium channels (ROCC), and store-operated channels (SOCC), help to control the Ca2+ trafficking between cytoplasmic and extracellular environment. The observed Ca2+ flux in our RASFs is most probably due to the coupling of celastrol with SERCA (unpublished data) which further antagonizes the function of the receptor. The inactivation of SERCA stops the transportation of Ca2+ into the ER and results in cytosolic Ca2+ accumulation which mediates the expression of the validated genes. Upon celastrol treatment, CAST, LEPR, TOLLIP, and TRAP1, could be partially restored to the untreated level by BAPTA/AM implying that other unknown molecular mechanisms may underpin the regulatory machinery. After extensive literature review, the transcriptional regulations of CD40, TRAP1, NFATC3, and CAST, have been reported before which are related to cytosolic Ca2+ mobilization (Casanova et al., 2006; Landriscina et al., 2010), the remaining 19 genes appear to be newly discovered to have their expression machinery regulated by Ca2+ signaling.

The molecular pathways which are mediated by the products of our validated genes may form a complex network in RASFs with the constituting pathways crosstalk with each other. We accordingly generated a blueprint outlining such potential molecular circuitry using literature information documenting the signaling cascades that connect the products of individual validated gene to Ca2+ signaling, and RA pathogenesis. Although such signaling network is created by integrating the molecular pathways described in different cell types, they could potentially be involved in the regulatory machinery of RASFs which provided crucial hints for supporting follow-up functional experiments. Generally, these pathways could be summarized into five different categories of signaling cascade which are: (1) the calpains/calmodulin-calcineurin-NFATc3 axis; (2) PI3K-Akt/PKC-NF-κB axis; (3) JAK-Stat axis; (4) ERK-MMP axis; and (5) MyD88-AP1 axis. The calpains/calmodulin-calcineurin-NFATc3 axis is per se a well-known Ca2+-associated pathway inevitably regulating the cartilage catabolism and osteoclast formation, angiogenesis, and inflammation during RA pathogenesis (Sitara and Aliprantis, 2010). Upon Ca2+ influx, the calcium sensor proteins, including calpains and calmodulin, bind to the cation and activate calcineurin which further dephosphorylates NFATc3 (Im and Rao, 2004). After nuclear translocation, NFATc3 complexes with other cell-type specific transcription factors (TF) for regulating the downstream expression of genes, like RANKL, which is the molecular culprit causing bone erosion (Wu et al., 2007). Because of the capacity of NFATc3 to partner with other TF in various tissues or cellular components, the calpains/calmodulin-calcineurin-NFATc3-mediated gene expression could lead to a widespread of pathogenic effects. Our validated genes also include calpastatin which is the upstream inhibitor for calpains suggesting that cellular Ca2+ is holistically involving in the regulation of the various signaling pathways. Therefore, targeting the calpains/calmodulin-calcineurin-NFATc3 axis via the regulation of cytosolic Ca2+ distribution with Ca2+-mobilizing agent like celastrol, is a potential strategy for intervening RA progression. It is worth noting that, calpains could be the central hub in our molecular circuitry diagram intertwining the various signaling cascades. For example, calpains can intervene with the JAK-STAT axis by inhibiting the activity of suppressor of cytokine signaling 3 (SOCS3) (Pothlichet et al., 2008; Miyazaki et al., 2015). For the PI3K-Akt/PKC-NF-κB axis, calpains can regulate the pathway by inhibiting at the level upstream of NF-κB (Li et al., 2011, 2014; Storr et al., 2011, 2015). Also, calpains can manipulate the ERK-MMP axis by direct activation of ERK1/2 (Moshal et al., 2006). Although cellular and animal test are needed for further validation of our findings, other studies using AIA rat showed that celastrol can modulate the NF-κB pathway, MAPK pathway, and the JAK/STAT pathway (Venkatesha et al., 2016) which may support the signaling circuitry as proposed in this report. Also, the PI3K-Akt/PKC-NF-κB axis may significantly be regulated by celastrol-induced Ca2+ flux in RASF, since more than one third of the validated genes, including histamine receptor H1 (HRH1), CD40, NFKB1, LEPR, IL4R, FGF10, and ERBB2, potentially target this signaling axis with most of them functioned molecularly via PI3K, the upstream kinase of Akt and PKC. NF-κB involved extensively in the development of chronic inflammation (Tak and Firestein, 2001), our findings point toward the efficacy of controlling RA progression by regulating the balance of cellular Ca2+. On the other hand, the pathway mediating the toll-like receptor and IL-1α/βsignaling are also the potential target of the genes TOLLIP, TLR6, and IL1R1 via the MyD88-AP1 axis (Dower et al., 1986; Burns et al., 1998, 2000; Bulut et al., 2001; Ahmad et al., 2007; de Almeida et al., 2013). The manipulation of such genes could, therefore, regulate the innate immune responses such as cellular migration and inflammation. There are several validated genes of which the corresponding signaling pathways cannot be well defined or lack of information acquired from the literature search. However, the products of these genes are known to participate in the development of inflammation like invasion, angiogenesis, cellular proliferation, and etc. The functions of these genes (ADGRE5, BMP1, CMTM1, IK, NFX1, and SCUBE1) and the associated molecular mechanisms underlying the corresponding signaling, therefore, deserve in-depth investigation for completing the signaling network as depicted in this study.

Taken together, we have discovered that cellular Ca2+ homeostasis is an important factor regulating the expression of a group of genes in RASFs which may significantly affect RA pathogenesis. As supported by the bioinformatics analysis, this Ca2+-dependent mechanism induced by celastrol is a new mode of action which has not been documented previously by using other conventional RA pharmaceutical interventions such as methotrexate, rituximab, and tocilizumab (Burmester and Pope, 2017; McInnes and Schett, 2017). However, the use of celastrol also demonstrated similarities to two out of three of the examined FDA-approved RA drugs (methotrexate and tocilizumab) in terms of the expression of some genes for example BMP1 and ERBB2 which support the therapeutic potency of celastrol in RA treatment. In addition, products of the celastrol induced Ca2+-mediated genes are participating in both upstream and downstream of molecular mechanisms leading to a comprehensive signaling control in a top-down basis not just targeting a single signaling messenger within a particular pathway. Therefore, our data highlighted the potential application of compounds, like celastrol, which are capable of regulating cytosolic Ca2+ level, as molecular tools for investigating the pathomechanisms of RA and other chronic inflammatory disorders. Our findings also provide forecasting platform for the establishment of functional experiments to validate our recent findings by using in vitro and in vivo models which facilitate the exploitation of novel pharmaceutical targets and therapeutic compounds.

Author Contributions

IdSRD and SM: conducted the experiments and drafted the manuscript. FG-M: prepared the figures and, materials and methods. IK and WH: conducted the bioinformatics analysis. BL: prepared the discussion and revised the whole manuscript. VW and LL: conceived the idea and designed the experimental plan.

Funding

This work was supported by a FDCT grant from the Macao Science and Technology Development Fund (Project code: 084/2013/A3).

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Acknowledgments

The authors would like to thank Prof. Erwin Neher for his critical review and comment of the manuscript.

Supplementary Material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fphar.2017.00962/full#supplementary-material

Abbreviations

RA, Rheumatoid arthritis; RASF, rheumatoid arthritis synovial fibroblasts; PCR, polymerase chain reaction; ADGRE5, adhesion G protein-coupled receptor E5; BMP1, bone morphogenetic protein 1; CAST, calpastatin; CD40, cluster of differentiation 40; CMTM1, CKLF-like MARVEL transmembrane domain containing 1; ERBB2, Erb-B2 receptor tyrosine kinase 2; FGF10, fibroblast growth factor 10; GLMN, glomulin; HRH1, histamine receptor H1; IFNAR1, interferon α and β receptor subunit 1; IK, interferon inhibiting cytokine factor; IL1R1, interleukin 1 receptor type 1; IL4R, interleukin 4 receptor; LEPR, leptin receptor; NFATC3, nuclear factor of activated T-cells 3; NFKB1, nuclear factor κB subunit 1; NFRKB, nuclear factor related to κB binding protein; NFX1, nuclear transcription factor, X-box binding 1; SCUBE1, signal peptide, CUB domain, EGF-like domain-containing protein 1; STAT3, signal transducer and activator of transcription 3; TLR6, toll-like receptor 6; TOLLIP, toll interacting protein; TRAP1, TNF receptor-associated protein 1.

References

Ahmad, R., Sylvester, J., and Zafarullah, M. (2007). MyD88, IRAK1 and TRAF6 knockdown in human chondrocytes inhibits interleukin-1-induced matrix metalloproteinase-13 gene expression and promoter activity by impairing MAP kinase activation. Cell. Signal. 19, 2549–2557. doi: 10.1016/j.cellsig.2007.08.013

PubMed Abstract | CrossRef Full Text | Google Scholar

Ai, M., Qiu, S., Lu, Y., and Fan, Z. (2013). HER2 regulates Brk/PTK6 stability via upregulating calpastatin, an inhibitor of calpain. Cell. Signal. 25, 1754–1761. doi: 10.1016/j.cellsig.2013.05.010

PubMed Abstract | CrossRef Full Text | Google Scholar

Albers, J. M., Kuper, H. H., van Riel, P. L., Prevoo, M. L., van 't Hof, M. A., van Gestel, A. M., et al. (1999). Socio-economic consequences of rheumatoid arthritis in the first years of the disease. Rheumatology 38, 423–430. doi: 10.1093/rheumatology/38.5.423

PubMed Abstract | CrossRef Full Text | Google Scholar

Arnett, F. C., Edworthy, S. M., Bloch, D. A., McShane, D. J., Fries, J. F., Cooper, N. S., et al. (1988). The American Rheumatism Association 1987 revised criteria for the classification of rheumatoid arthritis. Arthritis Rheum. 31, 315–324. doi: 10.1002/art.1780310302

PubMed Abstract | CrossRef Full Text | Google Scholar

Audard, V., Pawlak, A., Candelier, M., Lang, P., and Sahali, D. (2012). Upregulation of nuclear factor-related kappa B suggests a disorder of transcriptional regulation in minimal change nephrotic syndrome. PLoS ONE 7:e30523. doi: 10.1371/journal.pone.0030523

PubMed Abstract | CrossRef Full Text | Google Scholar

Baron, V. T., Pio, R., Jia, Z., and Mercola, D. (2015). Early growth response 3 regulates genes of inflammation and directly activates IL6 and IL8 expression in prostate cancer. Br. J. Cancer 112, 755–764. doi: 10.1038/bjc.2014.622

PubMed Abstract | CrossRef Full Text | Google Scholar

Bartok, B., and Firestein, G. S. (2010). Fibroblast-like synoviocytes: key effector cells in rheumatoid arthritis. Immunol. Rev. 233, 233–255. doi: 10.1111/j.0105-2896.2009.00859.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Brinker, A. M., Ma, J., Lipsky, P. E., and Raskin, I. (2007). Medicinal chemistry and pharmacology of genus Tripterygium (Celastraceae). Phytochemistry 68, 732–766. doi: 10.1016/j.phytochem.2006.11.029

PubMed Abstract | CrossRef Full Text | Google Scholar

Brun, M., and Godbout, R. (2016). Activation of calcineurin in cancer: many paths, one hub. Trans. Cancer Res. 5(Suppl. 3), S497–S506. doi: 10.21037/tcr.2016.09.30

CrossRef Full Text | Google Scholar

Bulut, Y., Faure, E., Thomas, L., Equils, O., and Arditi, M. (2001). Cooperation of Toll-like receptor 2 and 6 for cellular activation by soluble tuberculosis factor and Borrelia burgdorferi outer surface protein A lipoprotein: role of Toll-interacting protein and IL-1 receptor signaling molecules in Toll-like receptor 2 signaling. J. Immunol. 167, 987–994. doi: 10.4049/jimmunol.167.2.987

PubMed Abstract | CrossRef Full Text | Google Scholar

Burmester, G. R., and Pope, J. E. (2017). Novel treatment strategies in rheumatoid arthritis. Lancet 389, 2338–2348. doi: 10.1016/S0140-6736(17)31491-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Burns, K., Clatworthy, J., Martin, L., Martinon, F., Plumpton, C., Maschera, B., et al. (2000). Tollip, a new component of the IL-1RI pathway, links IRAK to the IL-1 receptor. Nat. Cell Biol. 2, 346–351. doi: 10.1038/35014038

PubMed Abstract | CrossRef Full Text | Google Scholar

Burns, K., Martinon, F., Esslinger, C., Pahl, H., Schneider, P., Bodmer, J. L., et al. (1998). MyD88, an adapter protein involved in interleukin-1 signaling. J. Biol. Chem. 273, 12203–12209. doi: 10.1074/jbc.273.20.12203

PubMed Abstract | CrossRef Full Text | Google Scholar

Casanova, M., Furlán, C., Sterin-Borda, L., and Borda, E. S. (2006). Muscarinic cholinoceptor activation modulates DNA synthesis and CD40 expression in fibroblast cells. Auton. Autacoid Pharmacol. 26, 293–301. doi: 10.1111/j.1474-8673.2006.00369.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Cascão, R., Vidal, B., Lopes, I. P., Paisana, E., Rino, J., Moita, L. F., et al. (2015). Decrease of CD68 Synovial macrophages in celastrol treated arthritic rats. PLoS ONE 10:e0142448. doi: 10.1371/journal.pone.0142448

PubMed Abstract | CrossRef Full Text | Google Scholar

Cascão, R., Vidal, B., Raquel, H., Neves-Costa, A., Figueiredo, N., Gupta, V., et al. (2012). Effective treatment of rat adjuvant-induced arthritis by celastrol. Autoimmun. Rev. 11, 856–862. doi: 10.1016/j.autrev.2012.02.022

PubMed Abstract | CrossRef Full Text | Google Scholar

Clapham, D. E. (2007). Calcium signaling. Cell 131, 1047–1058. doi: 10.1016/j.cell.2007.11.028

PubMed Abstract | CrossRef Full Text | Google Scholar

Dai, Y. S., Xu, J., and Molkentin, J. D. (2005). The DnaJ-related factor Mrj interacts with nuclear factor of activated T cells c3 and mediates transcriptional repression through class II histone deacetylase recruitment. Mol. Cell. Biol. 25, 9936–9948. doi: 10.1128/MCB.25.22.9936-9948.2005

PubMed Abstract | CrossRef Full Text | Google Scholar

de Almeida, L. A., Macedo, G. C., Marinho, F. A., Gomes, M. T., Corsetti, P. P., Silva, A. M., et al. (2013). Toll-like receptor 6 plays an important role in host innate resistance to Brucella abortus infection in mice. Infect. Immun. 81, 1654–1662. doi: 10.1128/IAI.01356-12

PubMed Abstract | CrossRef Full Text | Google Scholar

Dickenson, J. M. (2002). Stimulation of protein kinase B and p70 S6 kinase by the histamine H1 receptor in DDT1MF-2 smooth muscle cells. Br. J. Pharmacol. 135, 1967–1976. doi: 10.1038/sj.bjp.0704664

PubMed Abstract | CrossRef Full Text | Google Scholar

Dower, S. K., Kronheim, S. R., Hopp, T. P., Cantrell, M., Deeley, M., Gillis, S., et al. (1986). The cell surface receptors for interleukin-1 alpha and interleukin-1 beta are identical. Nature 324, 266–268. doi: 10.1038/324266a0

PubMed Abstract | CrossRef Full Text | Google Scholar

Dubois, G. R., Schweizer, R. C., Versluis, C., Bruijnzeel-Koomen, C. A., and Bruijnzeel, P. L. (1998). Human eosinophils constitutively express a functional interleukin-4 receptor: interleukin-4 -induced priming of chemotactic responses and induction of PI-3 kinase activity. Am. J. Respir. Cell Mol. Biol. 19, 691–699. doi: 10.1165/ajrcmb.19.4.3208

PubMed Abstract | CrossRef Full Text | Google Scholar

Firestein, G. S. (2003). Evolving concepts of rheumatoid arthritis. Nature 423, 356–361. doi: 10.1038/nature01661

PubMed Abstract | CrossRef Full Text | Google Scholar

Hanna, R. A., Campbell, R. L., and Davies, P. L. (2008). Calcium-bound structure of calpain and its mechanism of inhibition by calpastatin. Nature 456, 409–412. doi: 10.1038/nature07451

PubMed Abstract | CrossRef Full Text | Google Scholar

Hernández, G. L., Volpert, O. V., Iñiguez, M. A., Lorenzo, E., Martínez-Martínez, S., Grau, R., et al. (2001). Selective inhibition of vascular endothelial growth factor-mediated angiogenesis by cyclosporin A: roles of the nuclear factor of activated T cells and cyclooxygenase 2. J. Exp. Med. 193, 607–620. doi: 10.1084/jem.193.5.607

PubMed Abstract | CrossRef Full Text | Google Scholar

Im, S. H., and Rao, A. (2004). Activation and deactivation of gene expression by Ca2+/calcineurin-NFAT-mediated signaling. Mol. Cells 18, 1–9.

PubMed Abstract | Google Scholar

Ishikawa, H., Nakagawa, Y., Shimizu, K., Nishihara, H., Matsusue, Y., and Nakamura, T. (1999). Inflammatory cytokines induced down-regulation of m-calpain mRNA expression in fibroblastic synoviocytes from patients with osteoarthritis and rheumatoid arthritis. Biochem. Biophys. Res. Commun. 266, 341–346. doi: 10.1006/bbrc.1999.1819

PubMed Abstract | CrossRef Full Text | Google Scholar

Izquierdo, J. H., Bonilla-Abadía, F., Cañas, C. A., and Tobón, G. J. (2014). Calcium, channels, intracellular signaling and autoimmunity. Reumatol. Clin. 10, 43–47. doi: 10.1016/j.reuma.2013.05.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Jia, Y. Y., Lu, J., Huang, Y., Liu, G., Gao, P., Wan, Y. Z., et al. (2014). The involvement of NFAT transcriptional activity suppression in SIRT1-mediated inhibition of COX-2 expression induced by PMA/Ionomycin. PLoS ONE 9:e97999. doi: 10.1371/journal.pone.0097999

PubMed Abstract | CrossRef Full Text | Google Scholar

Kar, P., and Parekh, A. B. (2015). Distinct spatial Ca2+ signatures selectively activate different NFAT transcription factor isoforms. Mol. Cell 58, 232–243. doi: 10.1016/j.molcel.2015.02.027

PubMed Abstract | CrossRef Full Text | Google Scholar

Karin, M., and Ben-Neriah, Y. (2000). Phosphorylation meets ubiquitination: the control of NF-[kappa]B activity. Annu. Rev. Immunol. 18, 621–663. doi: 10.1146/annurev.immunol.18.1.621

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, D. H., Shin, E. K., Kim, Y. H., Lee, B. W., Jun, J. G., Park, J. H., et al. (2009). Suppression of inflammatory responses by celastrol, a quinone methide triterpenoid isolated from Celastrus regelii. Eur. J. Clin. Invest. 39, 819–827. doi: 10.1111/j.1365-2362.2009.02186.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Krummrei, U., Baulieu, E. E., and Chambraud, B. (2003). The FKBP-associated protein FAP48 is an antiproliferative molecule and a player in T cell activation that increases IL2 synthesis. Proc. Natl. Acad. Sci. U.S.A. 100, 2444–2449. doi: 10.1073/pnas.0438007100

PubMed Abstract | CrossRef Full Text | Google Scholar

Landriscina, M., Laudiero, G., Maddalena, F., Amoroso, M. R., Piscazzi, A., Cozzolino, F., et al. (2010). Mitochondrial chaperone Trap1 and the calcium binding protein Sorcin interact and protect cells against apoptosis induced by antiblastic agents. Cancer Res. 70, 6577–6586. doi: 10.1158/0008-5472.CAN-10-1256

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, H. L., Bae, O. Y., Baek, K. H., Kwon, A., Hwang, H. R., Qadir, A. S., et al. (2011). High extracellular calcium-induced NFATc3 regulates the expression of receptor activator of NF-kappaB ligand in osteoblasts. Bone 49, 242–249. doi: 10.1016/j.bone.2011.04.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, H. Y., Jeon, H. S., Song, E. K., Han, M. K., Park, S. I., Lee, S. I., et al. (2006). CD40 ligation of rheumatoid synovial fibroblasts regulates RANKL-mediated osteoclastogenesis: evidence of NF-kappaB-dependent, CD40-mediated bone destruction in rheumatoid arthritis. Arthr. Rheum. 54, 1747–1758. doi: 10.1002/art.21873

PubMed Abstract | CrossRef Full Text | Google Scholar

Lefèvre, S., Knedla, A., Tennie, C., Kampmann, A., Wunrau, C., Dinser, R., et al. (2009). Synovial fibroblasts spread rheumatoid arthritis to unaffected joints. Nat. Med. 15, 1414–1420. doi: 10.1038/nm.2050

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, X., Luo, R., Chen, R., Song, L., Zhang, S., Hua, W., et al. (2014). Cleavage of IkappaBalpha by calpain induces myocardial NF-kappaB activation, TNF-alpha expression, and cardiac dysfunction in septic mice. Am. J. Physiol. Heart Circ. Physiol. 306, H833–H843. doi: 10.1152/ajpheart.00893.2012

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, Y. H., Fu, H. L., Tian, M. L., Wang, Y. Q., Chen, W., Cai, L. L., et al. (2016). Neuron-derived FGF10 ameliorates cerebral ischemia injury via inhibiting NF-kappaB-dependent neuroinflammation and activating PI3K/Akt survival signaling pathway in mice. Sci. Rep. 6:19869. doi: 10.1038/srep19869

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, Y., Ma, J., Zhu, H., Singh, M., Hill, D., Greer, P. A., et al. (2011). Targeted inhibition of calpain reduces myocardial hypertrophy and fibrosis in mouse models of type 1 diabetes. Diabetes 60, 2985–2994. doi: 10.2337/db10-1333

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, J., Lee, J., Salazar Hernandez, M. A., Mazitschek, R., and Ozcan, U. (2015). Treatment of obesity with celastrol. Cell 161, 999–1011. doi: 10.1016/j.cell.2015.05.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Lv, Q. W., Zhang, W., Shi, Q., Zheng, W. J., Li, X., Chen, H., et al. (2015). Comparison of Tripterygium wilfordii Hook F with methotrexate in the treatment of active rheumatoid arthritis (TRIFRA): a randomised, controlled clinical trial. Ann. Rheum. Dis. 74, 1078–1086. doi: 10.1136/annrheumdis-2013-204807

PubMed Abstract | CrossRef Full Text | Google Scholar

Marchi, S., and Pinton, P. (2016). Alterations of calcium homeostasis in cancer cells. Curr. Opin. Pharmacol. 29, 1–6. doi: 10.1016/j.coph.2016.03.002

PubMed Abstract | CrossRef Full Text | Google Scholar

McInnes, I. B., and Schett, G. (2017). Pathogenetic insights from the treatment of rheumatoid arthritis. Lancet 389, 2328–2337. doi: 10.1016/S0140-6736(17)31472-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Minobe, E., Hao, L. Y., Saud, Z. A., Xu, J. J., Kameyama, A., Maki, M., et al. (2006). A region of calpastatin domain L that reprimes cardiac L-type Ca2+ channels. Biochem. Biophys. Res. Commun. 348, 288–294. doi: 10.1016/j.bbrc.2006.07.052

PubMed Abstract | CrossRef Full Text | Google Scholar

Miyazaki, T., Taketomi, Y., Saito, Y., Hosono, T., Lei, X. F., Kim-Kaneyama, J. R., et al. (2015). Calpastatin counteracts pathological angiogenesis by inhibiting suppressor of cytokine signaling 3 degradation in vascular endothelial cells. Circ. Res. 116, 1170–1181. doi: 10.1161/CIRCRESAHA.116.305363

PubMed Abstract | CrossRef Full Text | Google Scholar

Mognol, G. P., Carneiro, F. R., Robbs, B. K., Faget, D. V., and Viola, J. P. (2016). Cell cycle and apoptosis regulation by NFAT transcription factors: new roles for an old player. Cell Death Dis. 7:e2199. doi: 10.1038/cddis.2016.97

PubMed Abstract | CrossRef Full Text | Google Scholar

Mooren, F. C., and Kinne, R. K. (1998). Cellular calcium in health and disease. Biochim. Biophys. Acta 1406, 127–151. doi: 10.1016/S0925-4439(98)00006-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Moshal, K. S., Singh, M., Sen, U., Rosenberger, D. S., Henderson, B., Tyagi, N., et al. (2006). Homocysteine-mediated activation and mitochondrial translocation of calpain regulates MMP-9 in MVEC. Am. J. Physiol. Heart Circ. Physiol. 291, H2825–H2835. doi: 10.1152/ajpheart.00377.2006

PubMed Abstract | CrossRef Full Text | Google Scholar

Muller-Ladner, U., Ospelt, C., Gay, S., Distler, O., and Pap, T. (2007). Cells of the synovium in rheumatoid arthritis. Arthritis Res. Ther. 9:223. doi: 10.1186/ar2337

PubMed Abstract | CrossRef Full Text | Google Scholar

Nanjundaiah, S. M., Venkatesha, S. H., Yu, H., Tong, L., Stains, J. P., and Moudgil, K. D. (2012). Celastrus and its bioactive celastrol protect against bone damage in autoimmune arthritis by modulating osteoimmune cross-talk. J. Biol. Chem. 287, 22216–22226. doi: 10.1074/jbc.M112.356816

PubMed Abstract | CrossRef Full Text | Google Scholar

Neria, F., del Carmen Serrano-Perez, M., Velasco, P., Urso, K., Tranque, P., and Cano, E. (2013). NFATc3 promotes Ca(2+) -dependent MMP3 expression in astroglial cells. Glia 61, 1052–1066. doi: 10.1002/glia.22494

PubMed Abstract | CrossRef Full Text | Google Scholar

Niapour, M., Yu, Y., and Berger, S. A. (2008). Regulation of calpain activity by c-Myc through calpastatin and promotion of transformation in c-Myc-negative cells by calpastatin suppression. J. Biol. Chem. 283, 21371–21381. doi: 10.1074/jbc.M801462200

PubMed Abstract | CrossRef Full Text | Google Scholar

Ou, Y., Liu, L., Xue, L., Zhou, W., Zhao, Z., Xu, B., et al. (2014). TRAP1 shows clinical significance and promotes cellular migration and invasion through STAT3/MMP2 pathway in human esophageal squamous cell cancer. J. Genet. Genomics 41, 529–537. doi: 10.1016/j.jgg.2014.08.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Perlman, H., Liu, H., Georganas, C., Koch, A. E., Shamiyeh, E., Haines, G. K. III., et al. (2001). Differential expression pattern of the antiapoptotic proteins, Bcl-2 and FLIP, in experimental arthritis. Arthritis Rheum. 44, 2899–2908. doi: 10.1002/1529-0131(200112)44:12<2899::AID-ART478>3.0.CO;2-X

PubMed Abstract | CrossRef Full Text | Google Scholar

Pothlichet, J., Chignard, M., and Si-Tahar, M. (2008). Cutting edge: innate immune response triggered by influenza A virus is negatively regulated by SOCS1 and SOCS3 through a RIG-I/IFNAR1-dependent pathway. J. Immunol. 180, 2034–2038. doi: 10.4049/jimmunol.180.4.2034

PubMed Abstract | CrossRef Full Text | Google Scholar

Qian, J., Zheng, H., Huangfu, W. C., Liu, J., Carbone, C. J., Leu, N. A., et al. (2011). Pathogen recognition receptor signaling accelerates phosphorylation-dependent degradation of IFNAR1. PLoS Pathog. 7:e1002065. doi: 10.1371/journal.ppat.1002065

PubMed Abstract | CrossRef Full Text | Google Scholar

Rooke, R. (2014). Can calcium signaling be harnessed for cancer immunotherapy? Biochim. Biophys. Acta 1843, 2334–2340. doi: 10.1016/j.bbamcr.2014.01.034

PubMed Abstract | CrossRef Full Text | Google Scholar

Russell, A. S. (2008). Quality-of-life assessment in rheumatoid arthritis. Pharmacoeconomics 26, 831–846. doi: 10.2165/00019053-200826100-00004

PubMed Abstract | CrossRef Full Text | Google Scholar

Sanchez-Margalet, V., and Martin-Romero, C. (2001). Human leptin signaling in human peripheral blood mononuclear cells: activation of the JAK-STAT pathway. Cell. Immunol. 211, 30–36. doi: 10.1006/cimm.2001.1815

PubMed Abstract | CrossRef Full Text | Google Scholar

Schwaller, B. (2012). The regulation of a cell's Ca(2+) signaling toolkit: the Ca (2+) homeostasome. Adv. Exp. Med. Biol. 740, 1–25. doi: 10.1007/978-94-007-2888-2_1

PubMed Abstract | CrossRef Full Text | Google Scholar

Sitara, D., and Aliprantis, A. O. (2010). Transcriptional regulation of bone and joint remodeling by NFAT. Immunol. Rev. 233, 286–300. doi: 10.1111/j.0105-2896.2009.00849.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Sizemore, N., Leung, S., and Stark, G. R. (1999). Activation of phosphatidylinositol 3-kinase in response to interleukin-1 leads to phosphorylation and activation of the NF-kappaB p65/RelA subunit. Mol. Cell. Biol. 19, 4798–4805. doi: 10.1128/MCB.19.7.4798

PubMed Abstract | CrossRef Full Text | Google Scholar

Storr, S. J., Carragher, N. O., Frame, M. C., Parr, T., and Martin, S. G. (2011). The calpain system and cancer. Nat. Rev. Cancer 11, 364–374. doi: 10.1038/nrc3050

PubMed Abstract | CrossRef Full Text | Google Scholar

Storr, S. J., Thompson, N., Pu, X., Zhang, Y., and Martin, S. G. (2015). Calpain in breast cancer: role in disease progression and treatment response. Pathobiology 82, 133–141. doi: 10.1159/000430464

PubMed Abstract | CrossRef Full Text | Google Scholar

Tak, P. P., and Firestein, G. S. (2001). NF-kappaB: a key role in inflammatory diseases. J. Clin. Invest. 107, 7–11. doi: 10.1172/JCI11830

PubMed Abstract | CrossRef Full Text | Google Scholar

Tang, W., and Zuo, J. P. (2012). Immunosuppressant discovery from Tripterygium wilfordii Hook f: the novel triptolide analog (5R)-5-hydroxytriptolide (LLDT-8). Acta Pharmacol. Sin. 33, 1112–1118. doi: 10.1038/aps.2012.108

PubMed Abstract | CrossRef Full Text | Google Scholar

Tao, X., Younger, J., Fan, F. Z., Wang, B., and Lipsky, P. E. (2002). Benefit of an extract of Tripterygium Wilfordii Hook F in patients with rheumatoid arthritis: a double-blind, placebo-controlled study. Arthritis Rheum. 46, 1735–1743. doi: 10.1002/art.10411

PubMed Abstract | CrossRef Full Text | Google Scholar

Tarp, S., Eric Furst, D., Boers, M., Luta, G., Bliddal, H., Tarp, U., et al. (2017). Risk of serious adverse effects of biological and targeted drugs in patients with rheumatoid arthritis: a systematic review meta-analysis. Rheumatology 56, 417–425. doi: 10.1093/rheumatology/kew442

PubMed Abstract | CrossRef Full Text | Google Scholar

Uddin, S., Bavi, P., Siraj, A. K., Ahmed, M., Al-Rasheed, M., Hussain, A. R., et al. (2010). Leptin-R and its association with PI3K/AKT signaling pathway in papillary thyroid carcinoma. Endocr. Relat. Cancer 17, 191–202. doi: 10.1677/ERC-09-0153

PubMed Abstract | CrossRef Full Text | Google Scholar

Venkatesha, S. H., Astry, B., Nanjundaiah, S. M., Yu, H., and Moudgil, K. D. (2012). Suppression of autoimmune arthritis by Celastrus-derived Celastrol through modulation of pro-inflammatory chemokines. Bioorg. Med. Chem. 20, 5229–5234. doi: 10.1016/j.bmc.2012.06.050

PubMed Abstract | CrossRef Full Text | Google Scholar

Venkatesha, S. H., Dudics, S., Astry, B., and Moudgil, K. D. (2016). Control of autoimmune inflammation by celastrol, a natural triterpenoid. Pathog. Dis. 74:ftw059. doi: 10.1093/femspd/ftw059

PubMed Abstract | CrossRef Full Text | Google Scholar

Venkatesha, S. H., Yu, H., Rajaiah, R., Tong, L., and Moudgil, K. D. (2011). Celastrus-derived celastrol suppresses autoimmune arthritis by modulating antigen-induced cellular and humoral effector responses. J. Biol. Chem. 286, 15138–15146. doi: 10.1074/jbc.M111.226365

PubMed Abstract | CrossRef Full Text | Google Scholar

Walters, D. K., and Jelinek, D. F. (2004). A role for Janus kinases in crosstalk between ErbB3 and the interferon-alpha signaling complex in myeloma cells. Oncogene 23, 1197–1205. doi: 10.1038/sj.onc.1207203

PubMed Abstract | CrossRef Full Text | Google Scholar

Woods Ignatoski, K. M., Livant, D. L., Markwart, S., Grewal, N. K., and Ethier, S. P. (2003). The role of phosphatidylinositol 3'-kinase and its downstream signals in erbB-2-mediated transformation. Mol. Cancer Res. 1, 551–560.

PubMed Abstract | Google Scholar

Wu, H., Peisley, A., Graef, I. A., and Crabtree, G. R. (2007). NFAT signaling and the invention of vertebrates. Trends Cell Biol. 17, 251–260. doi: 10.1016/j.tcb.2007.04.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, C. H., Murti, A., Pfeffer, S. R., Fan, M., Du, Z., and Pfeffer, L. M. (2008). The role of TRAF2 binding to the type I interferon receptor in alternative NF kappaB activation and antiviral response. J. Biol. Chem. 283, 14309–14316. doi: 10.1074/jbc.M708895200

PubMed Abstract | CrossRef Full Text | Google Scholar

Yoon, H. Y., Lee, E. G., Lee, H., Cho, I. J., Choi, Y. J., Sung, M. S., et al. (2013). Kaempferol inhibits IL-1beta-induced proliferation of rheumatoid arthritis synovial fibroblasts and the production of COX-2, PGE2 and MMPs. Int. J. Mol. Med. 32, 971–977. doi: 10.3892/ijmm.2013.1468

PubMed Abstract | CrossRef Full Text | Google Scholar

Yoon, M. J., Lee, A. R., Jeong, S. A., Kim, Y. S., Kim, J. Y., Kwon, Y. J., et al. (2014). Release of Ca2+ from the endoplasmic reticulum and its subsequent influx into mitochondria trigger celastrol-induced paraptosis in cancer cells. Oncotarget 5, 6816–6831. doi: 10.18632/oncotarget.2256

PubMed Abstract | CrossRef Full Text | Google Scholar

Zenmyo, M., Hiraoka, K., Komiya, S., Morimatsu, M., and Sasaguri, Y. (1995). Histamine-stimulated production of matrix metalloproteinase 1 by human rheumatoid synovial fibroblasts is mediated by histamine H1-receptors. Virchows Arch. 427, 437–444. doi: 10.1007/BF00199394

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: Rheumatoid arthritis, RASFs, celastrol, calcium, inflammation, autoimmunity

Citation: de Seabra Rodrigues Dias IR, Mok SWF, Gordillo-Martínez F, Khan I, Hsiao WWL, Law BYK, Wong VKW and Liu L (2018) The Calcium-Induced Regulation in the Molecular and Transcriptional Circuitry of Human Inflammatory Response and Autoimmunity. Front. Pharmacol. 8:962. doi: 10.3389/fphar.2017.00962

Received: 30 August 2017; Accepted: 18 December 2017;
Published: 08 January 2018.

Edited by:

Rudolf Bauer, University of Graz, Austria

Reviewed by:

Linlin Lu, International Institute for Translational Chinese Medicine, China
Ouyang Chen, Second Military Medical University, China

Copyright © 2018 de Seabra Rodrigues Dias, Mok, Gordillo-Martínez, Khan, Hsiao, Law, Wong and Liu. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Vincent K. W. Wong, bowaiwong@gmail.com
Liang Liu, lliu@must.edu.mo

These authors have contributed equally to this work.

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.